1
|
Moghaddasi K, Hesaraki S, Arfaee F, Athari SS. Investigating the effect of mesenchymal stem cells on the rate of clinical and pathological improvement of asthmatic lung in mouse model. Regen Ther 2024; 25:157-161. [PMID: 38178929 PMCID: PMC10765480 DOI: 10.1016/j.reth.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/08/2023] [Accepted: 12/17/2023] [Indexed: 01/06/2024] Open
Abstract
Asthma is a pulmonary disease and its pathophysiology includes inflammation, obstruction, edema of the airways, and mucus secretions in the airways. Mesenchymal stem cells (MSCs) are self-renewal that use the therapeutic potential of these cells can be applied as treatments of asthma. In this study, the effect of Mesenchyme stem cells on asthma was investigated. MSCs were administrated for asthmatic mice and then, percentage of eosinophils in blood and bronchoalveolar lavage fluid (BALF), levels of interleukine (IL)-4 and Immunoglubolin (Ig)E were measured. Also histopathological study of lung tissue was done. MSCs administration could control percentage of eosinophils in blood and BALF, levels of IgE and IL-4, eosinophilic inflammation, mucin realizing and goblet cell hyper-plasia. Administration of MSCs as treatment of asthma can be a useful and applicable therapy in control of asthma symptoms.
Collapse
Affiliation(s)
- Kambiz Moghaddasi
- Department of Clinical Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Saeed Hesaraki
- Department of Clinical Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Farnoosh Arfaee
- Department of Clinical Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
2
|
Sun Y, Huang S, Liu K, Tang L, Liu X, Guo J, Zeng A, Ma Y, Li Z, Wang J, Su Y, Zhang P, Wang G, Guo W. Mesenchymal stem cells prevent H7N9 virus infection via rejuvenating immune environment to inhibit immune-overactivity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166973. [PMID: 38029943 DOI: 10.1016/j.bbadis.2023.166973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Influenza is a clinically important infectious disease with a high fatality rate, which always results in severe pneumonia. Mesenchymal stem cells (MSCs) exhibit promising therapeutic effects on severe viral pneumonia, but whether MSCs prevent virus infection and contribute to the prevention of influenza remains unknown. METHODS ICR mice were pretreated with human umbilical cord (hUC) MSCs and then infected with the influenza H7N9 virus. Weight, survival days, and lung index of mice were recorded. Serum antibody against influenza H7N9 virus was detected according to the hemagglutination inhibition method. Before and after virus infection, T cell and B cell subtypes in the peripheral blood of mice were evaluated by flow cytometry. Cytokines in the supernatants of MSCs, innate immune cells, and mouse broncho alveolar lavage fluid (BALF) were determined by enzyme-linked immunosorbent assay (ELISA) or Luminex Assay. RESULTS Pretreatment with MSCs protected mice against influenza H7N9 virus infection. Weight loss, survival rate, and structural and functional damage to the lungs of infected mice were significantly improved. Mechanistically, MSCs modulated T lymphocyte response in virus-infected mice and inhibited the cGAS/STING pathway. Importantly, the protective effect of MSCs was mediated by cell-to-cell communications and attenuation of cytokine storm caused by immune overactivation.
Collapse
Affiliation(s)
- Yinhua Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, People's Republic of China
| | - Shihao Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, People's Republic of China
| | - Kaituo Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Lei Tang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, People's Republic of China
| | - Xiqing Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, People's Republic of China
| | - Jingtian Guo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, People's Republic of China
| | - Aizhong Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, People's Republic of China
| | - Yuxiao Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, People's Republic of China
| | - Zhuolan Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, People's Republic of China
| | - Jing Wang
- Jiangsu Renocell Biotech Co., Ltd., Nanjing, Jiangsu, People's Republic of China
| | - Yueyan Su
- Jiangsu Renocell Biotech Co., Ltd., Nanjing, Jiangsu, People's Republic of China
| | - Pinghu Zhang
- Institute of Translational Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China.
| | - Guangji Wang
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu, People's Republic of China; Jiangsu Renocell Biotech Co., Ltd., Nanjing, Jiangsu, People's Republic of China.
| | - Wei Guo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, People's Republic of China; Jiangsu Renocell Biotech Co., Ltd., Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
3
|
Zendedel E, Tayebi L, Nikbakht M, Hasanzadeh E, Asadpour S. Clinical Trials of Mesenchymal Stem Cells for the Treatment of COVID 19. Curr Stem Cell Res Ther 2024; 19:1055-1071. [PMID: 37815188 DOI: 10.2174/011574888x260032230925052240] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 10/11/2023]
Abstract
Mesenchymal Stem Cells (MSCs) are being investigated as a treatment for a novel viral disease owing to their immunomodulatory, anti-inflammatory, tissue repair and regeneration characteristics, however, the exact processes are unknown. MSC therapy was found to be effective in lowering immune system overactivation and increasing endogenous healing after SARS-CoV-2 infection by improving the pulmonary microenvironment. Many studies on mesenchymal stem cells have been undertaken concurrently, and we may help speed up the effectiveness of these studies by collecting and statistically analyzing data from them. Based on clinical trial information found on clinicaltrials. gov and on 16 November 2020, which includes 63 clinical trials in the field of patient treatment with COVID-19 using MSCs, according to the trend of increasing studies in this field, and with the help of meta-analysis studies, it is possible to hope that the promise of MSCs will one day be realized. The potential therapeutic applications of MSCs for COVID-19 are investigated in this study.
Collapse
Affiliation(s)
- Elham Zendedel
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Lobat Tayebi
- Marquett University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Mohammad Nikbakht
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Hasanzadeh
- Immunogenetics Research Center, Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shiva Asadpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
4
|
Kandula UR, Wake AD. Effectiveness of RCTs Pooling Evidence on Mesenchymal Stem Cell (MSC) Therapeutic Applications During COVID-19 Epidemic: A Systematic Review. Biologics 2023; 17:85-112. [PMID: 37223116 PMCID: PMC10202141 DOI: 10.2147/btt.s404421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023]
Abstract
Background Global pandemic identified as coronavirus disease 2019 (COVID-19) has resulted in a variety of clinical symptoms, from asymptomatic carriers to those with severe acute respiratory distress syndrome (SARS) and moderate upper respiratory tract symptoms (URTS). This systematic review aimed to determine effectiveness of stem cell (SC) applications among COVID-19 patients. Methods Multiple databases of PubMed, EMBASE, Science Direct, Google Scholar, Scopus, Web of Science, and Cochrane Library were used. Studies were screened, chosen, and included in this systematic review using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 flowchart diagram and PRISMA checklist. Included studies' quality was assessed employing Critical Appraisal Skills Programme (CASP) quality evaluation criteria for 14 randomized controlled trials (RCTs). Results Fourteen RCTs were performed between the years of 2020 to 2022, respectively, with a sample size n = 574 (treatment group (n = 318); control group (n = 256)) in multiple countries of Indonesia, Iran, Brazil, Turkey, China, Florida, UK, and France. The greatest sample size reported from China among 100 COVID-19 patients, while the lowest sample of 9 COVID-19 patients from Jakarta, Indonesia, and the patient's age ranges from 18 to 69 years. Studies applied to the type of SC were "Umbilical cord MSCs, MSCs secretome, MSCs, Placenta-derived MSCs, Human immature dental pulp SC, DW-MSC infusion, Wharton Jelly-derived MSCs". The injected therapeutic dose was 1 × 106 cells/kg, 1 × 107 cells/kg, 1 × 105 cells/kg, and 1 million cells/kg as per the evidence from the different studies. Studies focused on demographic variables, clinical symptoms, laboratory tests, Comorbidities, respiratory measures, concomitant therapies, Sequential Organ Failure Assessment score, mechanical ventilation, body mass index, adverse events, inflammatory markers, and PaO2/FiO2 ratio were all recorded as study characteristics. Conclusion Clinical evidence on MSC's therapeutic applications during COVID-19 pandemic has proven to be a promising therapy for COVID-19 patient recovery with no consequences and applied as a routine treatment for challenging ailments.
Collapse
Affiliation(s)
- Usha Rani Kandula
- Department of Clinical Nursing, College of Health Sciences, Arsi University, Asella, Ethiopia
| | - Addisu Dabi Wake
- Department of Clinical Nursing, College of Health Sciences, Arsi University, Asella, Ethiopia
| |
Collapse
|
5
|
Pordanjani PM, Bolhassani A, Milani A, Pouriayevali MH. Extracellular vesicles in vaccine development and therapeutic approaches for viral diseases. Process Biochem 2023. [DOI: 10.1016/j.procbio.2023.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
|
6
|
Human Umbilical Cord Mesenchymal Stem Cells Attenuate Severe Burn-Induced Multiple Organ Injury via Potentiating IGF-1 and BCL-2/BAX Pathway. Stem Cells Int 2022; 2022:5474289. [PMID: 36591374 PMCID: PMC9803581 DOI: 10.1155/2022/5474289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/15/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022] Open
Abstract
Background Early multiple organ injuries induced by severe burn predict a high mortality. Mesenchymal stem cells (MSCs) are able to repair and reconstruct the injured tissues and organs induced by trauma and diseases. However, potential protective effect and mechanism of MSCs on multiorgan injury induced by severe burn at early stage remain to be not clarified. Therefore, this study was to explore the effect and mechanism of human umbilical cord-derived MSCs (hUCMSCs) against severe burn-induced early organ injuries in rats. Methods Adult male Wistar rats were randomly divided into sham, burn, and burn+hUCMSCsgroups. GFP-labeled hUCMSCs or PBS was intravenous injected into respective groups. Migration and distribution patterns of GFP-labeled hUCMSCs were observed by inverted fluorescence microscope. The structures and cell apoptosis of the heart, kidney, and liver were measured by immunohistochemistry. Biochemical parameters in serum were assayed by standard Roche-Hitachi methodology. Western blotting was performed on these organs of rats in the three groups to explore the underlying mechanisms. Results At 24 hours after hUCMSCs transplantation, we found that GFP-labeled hUCMSCs mainly localized in the blood vessel of the heart, kidney, and liver and a very few cells migrated into tissues of these organs. Compared with the sham group, structure damages and cell apoptosis of these organs were induced by severe burn, and systematic administrations of hUCMSCs significantly improved the damaged structures, cell apoptosis rates, and biochemical parameters of these organs. Furthermore, IGF-1 (insulin-like growth factor 1) level in burn+hUCMSCs group was significantly higher than that in the sham and burn groups. Meanwhile, severe burn induced BCL-2/BAX significantly decreased compared to the sham group, and it was markedly increased by hUCMSCs administration. Conclusion The hUCMSCs transplantation can attenuate severe burn-induced early organ injuries and protect multiorgan functions by encouraging migration of hUCMSCs with blood circulation and increasing protective cytokine IGF-1 level and regulating BCL-2/BAX pathway of these vital organs. Furthermore, these data might provide the theoretical foundation for further clinical applications of hUCMSCs in burn areas.
Collapse
|
7
|
Fujita Y, Nishimura M, Wada T, Komori N, Otoi T. Dimethyl sulfoxide-free cryopreservation solution containing trehalose, dextran 40, and propylene glycol for therapy with human adipose tissue-derived mesenchymal stromal cells. Cytotechnology 2022; 74:515-529. [PMID: 36238270 PMCID: PMC9525547 DOI: 10.1007/s10616-022-00541-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/24/2022] [Indexed: 01/22/2023] Open
Abstract
We evaluated a dimethyl sulfoxide (Me2SO)-free cryopreservation solution to freeze human adipose-derived mesenchymal stromal cells (hADSCs). In the first experiment, we compared the combined effects of 3% trehalose (3 T) and 5% dextran (5D) in lactated Ringer’s solution (LR) as a cryopreservation base solution containing 10% propylene glycol (PG). The cell viability of hADSCs immediately after thawing was significantly higher (p < 0.05) in LR supplemented with 3 T (LR-3 T) and with 3 T and 5D (LR-3 T-5D) than in LR. In the second experiment, we compared the cell characteristics of hADSCs freeze-thawed in LR-3 T-5D containing either 10% Me2SO or 10% PG. The cell viability, annexin V-positive ratio, colony-forming capacity, cell proliferation, cell surface antigen positivity, adipogenic differentiation, osteogenic differentiation, and genetic response to cytokine stimulation of hADSCs immediately after thawing were similar between the LR-3 T-5D containing 10% Me2SO and 10% PG. In the third experiment, we examined various concentrations of PG on the cell proliferative capacity of freeze-thawed hADSCs. The cell proliferative capacity of hADSCs frozen with LR-3 T-5D containing 2.5% to 5% PG was significantly higher (p < 0.05) than LR-3 T-5D containing 10% PG. Furthermore, the cell proliferative capacity of hADSCs frozen with LR-3 T-5D containing 4% PG was similar to that of fresh hADSCs. These results indicate that the combination of 3 T-5D in an LR solution as a basic solution is effective for post-thaw cell viability, and that the optimal concentration of PG to maintain the cell characteristics of hADSCs frozen with LR-3 T-5D is 2.5% to 5%, which is promising for cell therapy applications.
Collapse
Affiliation(s)
- Yasutaka Fujita
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc, Naruto, Tokushima 772-8601 Japan
| | - Masuhiro Nishimura
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc, Naruto, Tokushima 772-8601 Japan
| | - Tamaki Wada
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc, Naruto, Tokushima 772-8601 Japan
| | - Natsuki Komori
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc, Naruto, Tokushima 772-8601 Japan
| | - Takeshige Otoi
- Bio-Innovation Research Center, Tokushima University, 2272-2 Ishii, Myozai-gun, Tokushima 779-3233 Japan
| |
Collapse
|
8
|
Pu D, Zhai X, Zhou Y, Xie Y, Tang L, Yin L, Liu H, Li L. A narrative review of COVID-19-related acute respiratory distress syndrome (CARDS): "typical" or "atypical" ARDS? ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:908. [PMID: 36111011 PMCID: PMC9469157 DOI: 10.21037/atm-22-3717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/08/2022] [Indexed: 11/06/2022]
Abstract
Background and Objective The coronavirus disease of 2019 (COVID-19) is highly infectious and mainly involves the respiratory system, with some patients rapidly progress to acute respiratory distress syndrome (ARDS), which is the leading cause of death in COVID-19 patients. Hence, fully understanding the features of COVID-19-related ARDS (CARDS) and early management of this disease would improve the prognosis and reduce the mortality of severe COVID-19. With the development of recent studies which have focused on CARDS, whether CARDS is "typical" or "atypical" ARDS has become a hotly debated topic. Methods We searched for relevant literature from 1999 to 2021 published in PubMed by using the following keywords and their combinations: "COVID-19", "CARDS", "ARDS", "pathophysiological mechanism", "clinical manifestations", "prognosis", and "clinical trials". Then, we analyzed, compared and highlighted the differences between classic ARDS and CARDS from all of the aspects above. Key Content and Findings Classical ARDS commonly occurs within 1 week after a predisposing cause, yet the median time from symptoms onset to CARDS is longer than that of classical ARDS, manifesting within a period of 9.0-12.0 days. Although the lung mechanics exhibited in CARDS grossly match those of classical ARDS, there are some atypical manifestations of CARDS: the severity of hypoxemia seemed not to be proportional to injury of lung mechanics and an increase of thrombogenic processes. Meanwhile, some patients' symptoms do not correspond with the extent of the organic injury: a chest computed tomography (CT) will reveal the severe and diffuse lung injuries, yet the clinical presentations of patients can be mild. Conclusions Despite the differences between the CARDS and ARDS, in addition to the treatment of antivirals, clinicians should continue to follow the accepted evidence-based framework for managing all ARDS cases, including CARDS.
Collapse
Affiliation(s)
- Dan Pu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoqian Zhai
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuwen Zhou
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Xie
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Liansha Tang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Liyuan Yin
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hangtian Liu
- Data Science and Big Data Technology, Chengdu University of Information Technology, Chengdu, China
| | - Lu Li
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Human Placental Mesenchymal Stem Cells for the Treatment of ARDS in Rat. Stem Cells Int 2022; 2022:8418509. [PMID: 35756754 PMCID: PMC9226970 DOI: 10.1155/2022/8418509] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/21/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022] Open
Abstract
The acute respiratory distress syndrome (ARDS) is one of the main causes of high mortality in patients with coronavirus (COVID-19). In recent years, due to the coronavirus pandemic, the number of patients with ARDS has increased significantly. Unfortunately, until now, there are no effective treatments for ARDS caused by COVID-19. Many drugs are either ineffective or have a low effect. Currently, there have been reports of efficient use of mesenchymal stem cells (MSCs) for the treatment of ARDS caused by COVID-19. We investigated the influence of freeze-dried human placenta-derived mesenchymal stem cells (HPMSCs) in ARDS rat model. All animals have received intratracheal injection of 6 mg/kg of lipopolysaccharide (LPS). The rats were randomly divided into five groups: I: LPS, II: LPS+dexamethasone, III: LPS+HPMSCs, IV: HPMSC, and V: saline. ARDS observation time was short-term and amounted to 168 hours. The study has shown that HPMSCs are able to migrate and attach to damaged lung tissue, contributing to the resolution of pathology, restoration of function, and tissue repair in the alveolar space. Studies have also shown that the administration of HPMSCs in animals with ARDS model significantly reduced the levels of key cytokines such as IL-1β, IL-6, and TNF-α. Freeze-dried placental stem cell is a very promising biomaterial for the treatment of ARDS. The human placenta can be easily obtained because it is considered as a medical waste. At the same time, a huge number of MSCs can be obtained from the placental tissue, and there is no ethical controversy around their use. The freeze-dried MSCs from human placental tissue can be stored sterile at room temperature for a long time before use.
Collapse
|
10
|
Dave JR, Chandekar SS, Behera S, Desai KU, Salve PM, Sapkal NB, Mhaske ST, Dewle AM, Pokare PS, Page M, Jog A, Chivte PA, Srivastava RK, Tomar GB. Human gingival mesenchymal stem cells retain their growth and immunomodulatory characteristics independent of donor age. SCIENCE ADVANCES 2022; 8:eabm6504. [PMID: 35749495 PMCID: PMC9232118 DOI: 10.1126/sciadv.abm6504] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 05/06/2022] [Indexed: 05/28/2023]
Abstract
Aging has been reported to deteriorate the quantity and quality of mesenchymal stem cells (MSCs), which affect their therapeutic use in regenerative medicine. A dearth of age-related stem cell research further restricts their clinical applications. The present study explores the possibility of using MSCs derived from human gingival tissues (GMSCs) for studying their ex vivo growth characteristics and differentiation potential with respect to donor age. GMSCs displayed decreased in vitro adipogenesis and in vitro and in vivo osteogenesis with age, but in vitro neurogenesis remained unaffected. An increased expression of p53 and SIRT1 with donor age was correlated to their ability of eliminating tumorigenic events through apoptosis or autophagy, respectively. Irrespective of donor age, GMSCs displayed effective immunoregulation and regenerative potential in a mouse model of LPS-induced acute lung injury. Thus, we suggest the potential of GMSCs for designing cell-based immunomodulatory therapeutic approaches and their further extrapolation for acute inflammatory conditions such as acute respiratory distress syndrome and COVID-19.
Collapse
Affiliation(s)
- Jay R. Dave
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Sayali S. Chandekar
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Shubhanath Behera
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, 411007 Maharashtra, India
| | - Kaushik U. Desai
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Pradnya M. Salve
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Neha B. Sapkal
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Suhas T. Mhaske
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Ankush M. Dewle
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Parag S. Pokare
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Megha Page
- Department of Dentistry, Deenanath Mangeshkar Hospital and Research Centre, Pune, 411004 Maharashtra, India
| | - Ajay Jog
- Department of Dentistry, Deenanath Mangeshkar Hospital and Research Centre, Pune, 411004 Maharashtra, India
| | - Pankaj A. Chivte
- Saraswati Danwantri Dental College and Hospital, Parbhani, 431401 Maharashtra, India
| | - Rupesh K. Srivastava
- Department of Biotechnology, All India Institute of Medical Science, New Delhi 110029, India
| | - Geetanjali B. Tomar
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| |
Collapse
|
11
|
Putra A, Widyatmoko A, Ibrahim S, Amansyah F, Amansyah F, Berlian MA, Retnaningsih R, Pasongka Z, Sari FE, Rachmad B. Case series of the first three severe COVID-19 patients treated with the secretome of hypoxia-mesenchymal stem cells in Indonesia. F1000Res 2022; 10:228. [PMID: 35350705 DOI: 10.12688/f1000research.51191.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2021] [Indexed: 01/31/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the outbreak of coronavirus disease 2019 (COVID-19), which has been rapidly spreading. Several guideline therapies have been proposed as a possible treatment for SARS-CoV-2, however, these therapies are not sufficient to treat a severe condition of SARS-CoV-2 infection characterised by the increase of D-dimer and C-reactive protein (CRP) levels, and patchy ground-glass opacities (GGOs). Secretome-mesenchymal stem cells (S-MSCs) produced by MSCs under hypoxia could excessively release several anti-inflammatory cytokines and growth factors to control the COVID-19 cytokine storm and accelerate lung injury improvement. This is the first study investigating the clinical outcomes of three severe COVID-19 patients admitted to the intensive care unit of three different hospitals in Indonesia treated with S-MSCs. The decrease of D-dimer and CRP level was reported for all patients treated with S-MSCs. This was in line with improvement of pulmonary radiology, blood gas level, and hematologic assessment. In conclusion, these cases suggest that S-MSCs could effectively control D-dimer, CRP level and GGOs of severe COVID-19 patients associated with recovered pulmonary function.
Collapse
Affiliation(s)
- Agung Putra
- Stem Cell and Cancer Research (SCCR), Faculty of Medicine, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia.,Department of Postgraduate Biomedical Science, Faculty of Medicine, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia.,Department of Pathological Anatomy, Faculty of Medicine,, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia
| | - Agus Widyatmoko
- Department of Internal Medicine, Faculty of Medicine, Universitas Muhammadiyah Yogyakarta, Yogyakarta, Yogyakarta, Indonesia
| | - Sugeng Ibrahim
- Stem Cell and Cancer Research (SCCR), Faculty of Medicine, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia.,Department of Molecular Biology, Faculty of Medicine, Universitas Katholik (Unika) Soegijapranata, Semarang, Central Java, Indonesia
| | - Fajar Amansyah
- Department of Internal Medicine, Bhayangkara Hospital, Makassar, South Sulawesi, Indonesia
| | - Farid Amansyah
- Department of Internal Medicine, Bhayangkara Hospital, Makassar, South Sulawesi, Indonesia
| | - Mukti Arja Berlian
- Department of Internal Medicine, Dr. Esnawan Antariksa Air Force Hospital, Jakarta, Jakarta, Indonesia
| | - Retnaningsih Retnaningsih
- Department of Neurology and Intensive Care Unit, Kariadi Hospital, Universitas Diponegoro, Semarang, Central Java, Indonesia
| | | | - Flora Eka Sari
- Department of Pulmonary Medicine, Dr. Esnawan Antariksa Air Force Hospital, Jakarta, Jakarta, Indonesia
| | - Basuki Rachmad
- Department of Intensive Care Unit, Gatot Soebroto Army Hospital, Jakarta, Jakarta, Indonesia
| |
Collapse
|
12
|
Cellular therapies for the treatment and prevention of SARS-CoV-2 infection. Blood 2022; 140:208-221. [PMID: 35240679 PMCID: PMC8896869 DOI: 10.1182/blood.2021012249] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/01/2022] [Indexed: 12/15/2022] Open
Abstract
Patients with blood disorders who are immune suppressed are at increased risk for infection with severe acute respiratory syndrome coronavirus 2. Sequelae of infection can include severe respiratory disease and/or prolonged duration of viral shedding. Cellular therapies may protect these vulnerable patients by providing antiviral cellular immunity and/or immune modulation. In this recent review of the field, phase 1/2 trials evaluating adoptive cellular therapies with virus-specific T cells or natural killer cells are described along with trials evaluating the safety, feasibility, and preliminary efficacy of immune modulating cellular therapies including regulatory T cells and mesenchymal stromal cells. In addition, the immunologic basis for these therapies is discussed.
Collapse
|
13
|
Kebria MM, Milan PB, Peyravian N, Kiani J, Khatibi S, Mozafari M. Stem cell therapy for COVID-19 pneumonia. MOLECULAR BIOMEDICINE 2022; 3:6. [PMID: 35174448 PMCID: PMC8850486 DOI: 10.1186/s43556-021-00067-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 12/22/2021] [Indexed: 12/11/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) virus is a highly contagious microorganism, and despite substantial investigation, no progress has been achieved in treating post-COVID complications. However, the virus has made various mutations and has spread around the world. Researchers have tried different treatments to reduce the side effects of the COVID-19 symptoms. One of the most common and effective treatments now used is steroid therapy to reduce the complications of this disease. Long-term steroid therapy for chronic inflammation following COVID-19 is harmful and increases the risk of secondary infection, and effective treatment remains challenging owing to fibrosis and severe inflammation and infection. Sometimes our immune system can severely damage ourselves in disease. In the past, many researchers have conducted various studies on the immunomodulatory properties of stem cells. This property of stem cells led them to modulate the immune system of autoimmune diseases like diabetes, multiple sclerosis, and Parkinson's. Because of their immunomodulatory properties, stem cell-based therapy employing mesenchymal or hematopoietic stem cells may be a viable alternative treatment option in some patients. By priming the immune system and providing cytokines, chemokines, and growth factors, stem cells can be employed to build a long-term regenerative and protective response. This review addresses the latest trends and rapid progress in stem cell treatment for Acute Respiratory Distress Syndrome (ARDS) following COVID-19.
Collapse
Affiliation(s)
- Maziar Malekzadeh Kebria
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Peiman Brouki Milan
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Noshad Peyravian
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Present Address: Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soheil Khatibi
- Babol University of Medical Sciences, Infection Diseases Centre, Mazandaran, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Schultz IC, Bertoni APS, Wink MR. Purinergic signaling elements are correlated with coagulation players in peripheral blood and leukocyte samples from COVID-19 patients. J Mol Med (Berl) 2022; 100:569-584. [PMID: 35091759 PMCID: PMC8799442 DOI: 10.1007/s00109-021-02175-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 12/20/2022]
Abstract
For over a year, the coronavirus disease 2019 has been affecting the world population by causing severe tissue injuries and death in infected people. Adenosine triphosphate (ATP) and the nicotinamide adenine dinucleotide (NAD +) are two molecules that are released into the extracellular microenvironment after direct virus infection or cell death caused by hyper inflammation and coagulopathy. Also, these molecules are well known to participate in multiple pathways and have a pivotal role in the purinergic signaling pathway. Thus, using public datasets available on the Gene Expression Omnibus (GEO), we analyzed raw proteomics data acquired using mass spectrometry (the gold standard method) and raw genomics data from COVID-19 patient samples obtained by microarray. The data was analyzed using bioinformatics and statistical methods according to our objectives. Here, we compared the purinergic profile of the total leukocyte population and evaluated the levels of these soluble biomolecules in the blood, and their correlation with coagulation components in COVID-19 patients, in comparison to healthy people or non-COVID-19 patients. The blood metabolite analysis showed a stage-dependent inosine increase in COVID-19 patients, while the nucleotides ATP and ADP had positive correlations with fibrinogen and other coagulation proteins. Also, ATP, ADP, inosine, and hypoxanthine had positive and negative correlations with clinical features. Regarding leukocyte gene expression, COVID-19 patients showed an upregulation of the P2RX1, P2RX4, P2RX5, P2RX7, P2RY1, P2RY12, PANX1, ADORA2B, NLPR3, and F3 genes. Yet, the ectoenzymes of the canonical and non-canonical adenosinergic pathway (ENTPD1 and CD38) are upregulated, suggesting that adenosine is produced by both active adenosinergic pathways. Hence, approaches targeting these biomolecules or their specific purinoreceptors and ectoenzymes may attenuate the high inflammatory state and the coagulopathy seen in COVID-19 patients. KEY MESSAGES : Adenosinergic pathways are modulated on leukocytes from COVID-19 patients. Plasmatic inosine levels are increased in COVID-19 patients. ATP, ADP, AMP, hypoxanthine, and inosine are correlated with coagulation players. The nucleotides and nucleosides are correlated with patients' clinical features. The P2 receptors and ectoenzymes are correlated with Tissue factor in COVID-19.
Collapse
Affiliation(s)
- Iago C Schultz
- Departamento de Ciências Básicas da Saúde, Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245 Sala 304, Porto Alegre, RS, 90050-170, Brazil
| | - Ana Paula S Bertoni
- Departamento de Ciências Básicas da Saúde, Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245 Sala 304, Porto Alegre, RS, 90050-170, Brazil
| | - Márcia R Wink
- Departamento de Ciências Básicas da Saúde, Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245 Sala 304, Porto Alegre, RS, 90050-170, Brazil.
| |
Collapse
|
15
|
Ebrahimi M, Rad MTS, Zebardast A, Ayyasi M, Goodarzi G, Tehrani SS. The critical role of mesenchymal stromal/stem cell therapy in COVID-19 patients: An updated review. Cell Biochem Funct 2021; 39:945-954. [PMID: 34545605 PMCID: PMC8652792 DOI: 10.1002/cbf.3670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/02/2021] [Accepted: 09/04/2021] [Indexed: 12/20/2022]
Abstract
New coronavirus disease 2019 (COVID-19), as a pandemic disaster, has drawn the attention of researchers in various fields to discover suitable therapeutic approaches for the management of COVID-19 patients. Currently, there are many worries about the rapid spread of COVID-19; there is no approved treatment for this infectious disease, despite many efforts to develop therapeutic procedures for COVID-19. Emerging evidence shows that mesenchymal stromal/stem cell (MSC) therapy can be a suitable option for the management of COVID-19. These cells have many biological features (including the potential of differentiation, high safety and effectiveness, secretion of trophic factors and immunoregulatory features) that make them suitable for the treatment of various diseases. However, some studies have questioned the positive role of MSC therapy in the treatment of COVID-19. Accordingly, in this paper, we will focus on the therapeutic impacts of MSCs and their critical role in cytokine storm of COVID-19 patients.
Collapse
Affiliation(s)
- Mohsen Ebrahimi
- Neonatal and Child Health Research CenterGolestan University of Medical SciencesGorganIran
| | - Mohammad Taha Saadati Rad
- Psychiatric and Behavioral Sciences Research Center, Addiction Research InstituteMazandaran University of Medical SciencesSariIran
| | - Arghavan Zebardast
- Department of Virology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Mitra Ayyasi
- Critical Care NursingIslamic Azad University, Sari BranchSariIran
| | - Golnaz Goodarzi
- Department of Clinical Biochemistry, School of MedicineTehran University of Medical SciencesTehranIran
- Scientific Research CenterTehran University of Medical SciencesTehranIran
| | - Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of MedicineTehran University of Medical SciencesTehranIran
- Scientific Research CenterTehran University of Medical SciencesTehranIran
| |
Collapse
|
16
|
Salimi-Jeda A, Abbassi S, Mousavizadeh A, Esghaie M, Bokharaei-Salim F, Jeddi F, Shafaati M, Abdoli A. SARS-CoV-2: Current trends in emerging variants, pathogenesis, immune responses, potential therapeutic, and vaccine development strategies. Int Immunopharmacol 2021; 101:108232. [PMID: 34673335 PMCID: PMC8519814 DOI: 10.1016/j.intimp.2021.108232] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/18/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023]
Abstract
More than a year after the SARS-CoV-2 pandemic, the Coronavirus disease 19 (COVID-19) is still a major global challenge for scientists to understand the different dimensions of infection and find ways to prevent, treat, and develop a vaccine. On January 30, 2020, the world health organization (WHO) officially announced this new virus as an international health emergency. While many biological and mechanisms of pathogenicity of this virus are still unclear, it seems that cytokine storm resulting from an immune response against the virus is considered the main culprit of the severity of the disease. Despite many global efforts to control the SARS-CoV-2, several problems and challenges have been posed in controlling the COVID-19 infection. These problems include the various mutations, the emergence of variants with high transmissibility, the short period of immunity against the virus, the possibility of reinfection in people improved, lack of specific drugs, and problems in the development of highly sensitive and specific vaccines. In this review, we summarized the results of the current trend and the latest research studies on the characteristics of the structure and genome of the SARS-CoV- 2, new mutations and variants of SARS-CoV-2, pathogenicity, immune response, virus diagnostic tests, potential treatment, and vaccine candidate.
Collapse
Affiliation(s)
- Ali Salimi-Jeda
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Sina Abbassi
- Department of Anesthesiology, Faculty of Medical Science, Tehran University of Medical Science, Tehran, Iran
| | - Atieh Mousavizadeh
- Department of Virology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Maryam Esghaie
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farah Bokharaei-Salim
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farhad Jeddi
- Department of Medical Genetics and Pathology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Maryam Shafaati
- Department of Microbiology, Jahrom Branch, Islamic Azad University, Fars, Iran
| | - Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
17
|
The Potential of Mesenchymal Stem Cells for the Treatment of Cytokine Storm due to COVID-19. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3178796. [PMID: 34840969 PMCID: PMC8626179 DOI: 10.1155/2021/3178796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/24/2021] [Accepted: 10/29/2021] [Indexed: 12/15/2022]
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has seriously affected public health and social stability. The main route of the transmission is droplet transmission, where the oral cavity is the most important entry point to the body. Due to both the direct harmful effects of SARS-CoV-2 and disordered immune responses, some COVID-19 patients may progress to acute respiratory distress syndrome or even multiple organ failure. Genetic variants of SARS-CoV-2 have been emerging and circulating around the world. Currently, there is no internationally approved precise treatment for COVID-19. Mesenchymal stem cells (MSCs) can traffic and migrate towards the affected tissue, regulate both the innate and acquired immune systems, and participate in the process of healing. Here, we will discuss and investigate the mechanisms of immune disorder in COVID-19 and the therapeutic activity of MSCs, in particular human gingiva mesenchymal stem cells.
Collapse
|
18
|
Abbas M, Alqahtani MS, Almohiy HM, Alqahtani FF, Alhifzi R, Jambi LK. The Potential Contribution of Biopolymeric Particles in Lung Tissue Regeneration of COVID-19 Patients. Polymers (Basel) 2021; 13:4011. [PMID: 34833310 PMCID: PMC8623030 DOI: 10.3390/polym13224011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 02/08/2023] Open
Abstract
The lung is a vital organ that houses the alveoli, which is where gas exchange takes place. The COVID-19 illness attacks lung cells directly, creating significant inflammation and resulting in their inability to function. To return to the nature of their job, it may be essential to rejuvenate the afflicted lung cells. This is difficult because lung cells need a long time to rebuild and resume their function. Biopolymeric particles are the most effective means to transfer developing treatments to airway epithelial cells and then regenerate infected lung cells, which is one of the most significant symptoms connected with COVID-19. Delivering biocompatible and degradable natural biological materials, chemotherapeutic drugs, vaccines, proteins, antibodies, nucleic acids, and diagnostic agents are all examples of these molecules' usage. Furthermore, they are created by using several structural components, which allows them to effectively connect with these cells. We highlight their most recent uses in lung tissue regeneration in this review. These particles are classified into three groups: biopolymeric nanoparticles, biopolymeric stem cell materials, and biopolymeric scaffolds. The techniques and processes for regenerating lung tissue will be thoroughly explored.
Collapse
Affiliation(s)
- Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
- Computers and Communications Department, College of Engineering, Delta University for Science and Technology, Gamasa 35712, Egypt
| | - Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; (M.S.A.); (H.M.A.); (R.A.)
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, UK
| | - Hussain M. Almohiy
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; (M.S.A.); (H.M.A.); (R.A.)
| | - Fawaz F. Alqahtani
- Department of Radiological Sciences, College of Applied Medical Sciences, Najran University, Najran 1988, Saudi Arabia;
| | - Roaa Alhifzi
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; (M.S.A.); (H.M.A.); (R.A.)
| | - Layal K. Jambi
- Radiological Sciences Department, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia;
| |
Collapse
|
19
|
SARS-CoV-2 Exposed Mesenchymal Stromal Cell from Congenital Pulmonary Airway Malformations: Transcriptomic Analysis and the Expression of Immunomodulatory Genes. Int J Mol Sci 2021; 22:ijms222111814. [PMID: 34769246 PMCID: PMC8584055 DOI: 10.3390/ijms222111814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
The inflammatory response plays a central role in the complications of congenital pulmonary airway malformations (CPAM) and severe coronavirus disease 2019 (COVID-19). The aim of this study was to evaluate the transcriptional changes induced by SARS-CoV-2 exposure in pediatric MSCs derived from pediatric lung (MSCs-lung) and CPAM tissues (MSCs-CPAM) in order to elucidate potential pathways involved in SARS-CoV-2 infection in a condition of exacerbated inflammatory response. MSCs-lung and MSCs-CPAM do not express angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TRMPSS2). SARS-CoV-2 appears to be unable to replicate in MSCs-CPAM and MSCs-lung. MSCs-lung and MSCs-CPAM maintained the expression of stemness markers MSCs-lung show an inflammatory response (IL6, IL1B, CXCL8, and CXCL10), and the activation of Notch3 non-canonical pathway; this route appears silent in MSCs-CPAM, and cytokine genes expression is reduced. Decreased value of p21 in MSCs-lung suggested no cell cycle block, and cells did not undergo apoptosis. MSCs-lung appears to increase genes associated with immunomodulatory function but could contribute to inflammation, while MSCs-CPAM keeps stable or reduce the immunomodulatory receptors expression, but they also reduce their cytokines expression. These data indicated that, independently from their perilesional or cystic origin, the MSCs populations already present in a patient affected with CPAM are not permissive for SARS-CoV-2 entry, and they will not spread the disease in case of infection. Moreover, these MSCs will not undergo apoptosis when they come in contact with SARS-CoV-2; on the contrary, they maintain their staminality profile.
Collapse
|
20
|
Putra A, Widyatmoko A, Ibrahim S, Amansyah F, Amansyah F, Berlian MA, Retnaningsih R, Pasongka Z, Sari FE, Rachmad B. Case series of the first three severe COVID-19 patients treated with the secretome of hypoxia-mesenchymal stem cells in Indonesia. F1000Res 2021; 10:228. [PMID: 35350705 PMCID: PMC8927740 DOI: 10.12688/f1000research.51191.3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the outbreak of coronavirus disease 2019 (COVID-19), which has been rapidly spreading. Several guideline therapies have been proposed as a possible treatment for SARS-CoV-2, however, these therapies are not sufficient to treat a severe condition of SARS-CoV-2 infection characterised by the increase of D-dimer and C-reactive protein (CRP) levels, and patchy ground-glass opacities (GGOs). Secretome-mesenchymal stem cells (S-MSCs) produced by MSCs under hypoxia could excessively release several anti-inflammatory cytokines and growth factors to control the COVID-19 cytokine storm and accelerate lung injury improvement. This is the first study investigating the clinical outcomes of three severe COVID-19 patients admitted to the intensive care unit of three different hospitals in Indonesia treated with S-MSCs. The decrease of D-dimer and CRP level was reported for all patients treated with S-MSCs. This was in line with improvement of pulmonary radiology, blood gas level, and hematologic assessment. In conclusion, these cases suggest that S-MSCs could effectively control D-dimer, CRP level and GGOs of severe COVID-19 patients associated with recovered pulmonary function.
Collapse
Affiliation(s)
- Agung Putra
- Stem Cell and Cancer Research (SCCR), Faculty of Medicine, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia
- Department of Postgraduate Biomedical Science, Faculty of Medicine, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia
- Department of Pathological Anatomy, Faculty of Medicine,, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia
| | - Agus Widyatmoko
- Department of Internal Medicine, Faculty of Medicine, Universitas Muhammadiyah Yogyakarta, Yogyakarta, Yogyakarta, Indonesia
| | - Sugeng Ibrahim
- Stem Cell and Cancer Research (SCCR), Faculty of Medicine, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia
- Department of Molecular Biology, Faculty of Medicine, Universitas Katholik (Unika) Soegijapranata, Semarang, Central Java, Indonesia
| | - Fajar Amansyah
- Department of Internal Medicine, Bhayangkara Hospital, Makassar, South Sulawesi, Indonesia
| | - Farid Amansyah
- Department of Internal Medicine, Bhayangkara Hospital, Makassar, South Sulawesi, Indonesia
| | - Mukti Arja Berlian
- Department of Internal Medicine, Dr. Esnawan Antariksa Air Force Hospital, Jakarta, Jakarta, Indonesia
| | - Retnaningsih Retnaningsih
- Department of Neurology and Intensive Care Unit, Kariadi Hospital, Universitas Diponegoro, Semarang, Central Java, Indonesia
| | | | - Flora Eka Sari
- Department of Pulmonary Medicine, Dr. Esnawan Antariksa Air Force Hospital, Jakarta, Jakarta, Indonesia
| | - Basuki Rachmad
- Department of Intensive Care Unit, Gatot Soebroto Army Hospital, Jakarta, Jakarta, Indonesia
| |
Collapse
|
21
|
Najafloo R, Majidi J, Asghari A, Aleemardani M, Kamrava SK, Simorgh S, Seifalian A, Bagher Z, Seifalian AM. Mechanism of Anosmia Caused by Symptoms of COVID-19 and Emerging Treatments. ACS Chem Neurosci 2021; 12:3795-3805. [PMID: 34609841 PMCID: PMC8507153 DOI: 10.1021/acschemneuro.1c00477] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/20/2021] [Indexed: 01/08/2023] Open
Abstract
The occurrence of anosmia, the loss or change in sense of smell, is one of the most common symptoms of COVID-19 experienced by almost 53% of those affected. Several hypotheses explain the mechanism of anosmia in patients suffering from COVID-19. This study aims to review the related mechanisms and answer the questions regarding COVID-19-related anosmia as well as propose a new strategy for treatment of long-term anosmia as a result of COVID-19 infection. This paper covers all of the studies investigating olfactory disorders following COVID-19 infection and explains the possible reasons for the correlated anosmia, including olfactory cleft syndrome, local inflammation in the nasal epithelium, early apoptosis of olfactory cells, changes in olfactory cilia and odor transmission, damage to microglial cells, effect on olfactory bulbs, epithelial olfactory injury, and impairment of olfactory neurons and stem cells. The key questions that arise in this field have been discussed, such as why prevalent anosmia is varied among the age categories and among sexes and the correlation of anosmia with mild or severe COVID-19 infection. The angiotensin-converting enzyme 2 receptor is a significant player in the mechanism of anosmia in COVID-19 patients. Based on current studies, a novel approach to treat long-COVID-19 with ongoing anosmia has been proposed. The fields of smart drug delivery, tissue engineering, and cell therapy provide a hypothesized strategy that can minimize the side effects of current treatments and support efficient recovery of the olfactory system.
Collapse
Affiliation(s)
- Raziyeh Najafloo
- Department
of Tissue Engineering & Regenerative Medicine, Faculty of Advanced
Technologies in Medicine, Iran University
of Medical Sciences (IUMS), Tehran 1449614535, Iran
| | - Jila Majidi
- Department
of Tissue Engineering & Regenerative Medicine, Faculty of Advanced
Technologies in Medicine, Iran University
of Medical Sciences (IUMS), Tehran 1449614535, Iran
| | - Alimohamad Asghari
- Skull
Base Research Center, Hazrat Rasoul Akram Hospital, The Five Senses
Health Institute, Iran University of Medical
Sciences (IUMS), Tehran 1445613131, Iran
| | - Mina Aleemardani
- Biomaterials
and Tissue Engineering Group, Department of Materials Science and
Engineering, Kroto Research Institute, The
University of Sheffield, Sheffield S3 7HQ, United Kingdom
| | - Seyed Kamran Kamrava
- ENT
and Head and Neck Research Center and Department, Hazrat Rasoul Akram
Hospital, The Five Senses Health Institute, Iran University of Medical Sciences (IUMS), Tehran 1445613131, Iran
| | - Sara Simorgh
- Department
of Tissue Engineering & Regenerative Medicine, Faculty of Advanced
Technologies in Medicine, Iran University
of Medical Sciences (IUMS), Tehran 1449614535, Iran
| | - Amelia Seifalian
- University
College London Medical School (UCL), London WC1E 6BT, United
Kingdom
- Watford
General Hospital, Watford WD18 0HB, United Kingdom
| | - Zohreh Bagher
- Department
of Tissue Engineering & Regenerative Medicine, Faculty of Advanced
Technologies in Medicine, Iran University
of Medical Sciences (IUMS), Tehran 1449614535, Iran
- ENT
and Head and Neck Research Center and Department, Hazrat Rasoul Akram
Hospital, The Five Senses Health Institute, Iran University of Medical Sciences (IUMS), Tehran 1445613131, Iran
| | - Alexander M. Seifalian
- Nanotechnology
and Regenerative Medicine Commercialisation Centre (NanoRegMed Ltd.), London BioScience Innovation Centre, London NW1 0NH, United Kingdom
| |
Collapse
|
22
|
Montanucci P, Pescara T, Greco A, Francisci D, Basta G, Calafiore R. Microencapsulated Wharton Jelly-derived adult mesenchymal stem cells as a potential new therapeutic tool for patients with COVID-19 disease: an in vitro study. AMERICAN JOURNAL OF STEM CELLS 2021; 10:36-52. [PMID: 34552816 PMCID: PMC8449139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/07/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND The recent newly appeared Coronavirus disease (COVID-19), caused by an enveloped RNA virus named "severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)", is associated with severe respiratory morbidity and mortality. Recent studies have shown that lymphopenia and a cytokine mass release represent important pathogenic features, with clinical evidence of dyspnea and hypoxemia, often leading to acute respiratory distress syndrome (ARDS), in severely ill patients, with a high death toll. Currently, stem cells are actively being investigated for their potential use in many "untreatable" diseases. In this regard and in particular, Mesenchymal Stem Cells (MSC), due to their intrinsic features, including either ability to impact on regulation of the immune system, or association with both anti-viral and anti-inflammatory properties, or potential for differentiation into several cell lineages, have become a promising tool for cell and molecular-based therapies. On this background, we wished to explore whether human umbilical cord-derived mesenchymal stem cells (hUCMS) would represent a potential viable therapeutic approach for the management of critically ill COVID19 patients. METHODS We tested the hUCMS effects on peripheral blood mononuclear cell (PBMCs) retrieved from patients with COVID19 (Ethical Committee CEAS Umbria, Italy CER N°3658/20 7, May, 2020), both as free cell monolayers and after envelopment in sodium alginate microcapsules. Both cell systems, after priming with IFN-γ, proved able to produce several immunomodulatory molecules such as IDO1 and HLAG5, although only the microencapsulated hUCMS were associated with massive and dose-dependent production of these factors. RESULTS The microencapsulated hUCMS improved allo-suppression in mixed lymphocytes reactions (MLRs), while also blunting T helper 1 and T helper 17 responses, that are involved with the cytokine storm and greatly contribute to the patient death. Moreover, we observed that both free and microencapsulated hUCMS permitted 5 days survival of in vitro culture maintained PBMCs extracted from very ill patients. CONCLUSION We have provided evidence that microencapsulated hUCMS in vitro, seem to represent a powerful tool to impact on several immune pathways, clearly deranged in COVID19 patients. Further study is necessary to begin in vivo assessment of this experimental system, upon determining both, the most appropriate time of the disease onset for intervention, and cell dosage/patient of our experimental product.
Collapse
Affiliation(s)
- Pia Montanucci
- Division of Internal Medicine and Endocrine and Metabolic Sciences (MISEM), Laboratory for Endocrine Cell Transplants and Biohybrid Organs, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| | - Teresa Pescara
- Division of Internal Medicine and Endocrine and Metabolic Sciences (MISEM), Laboratory for Endocrine Cell Transplants and Biohybrid Organs, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| | - Alessia Greco
- Division of Internal Medicine and Endocrine and Metabolic Sciences (MISEM), Laboratory for Endocrine Cell Transplants and Biohybrid Organs, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| | - Daniela Francisci
- Division of Infectious Diseases, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| | - Giuseppe Basta
- Division of Internal Medicine and Endocrine and Metabolic Sciences (MISEM), Laboratory for Endocrine Cell Transplants and Biohybrid Organs, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| | - Riccardo Calafiore
- Division of Internal Medicine and Endocrine and Metabolic Sciences (MISEM), Laboratory for Endocrine Cell Transplants and Biohybrid Organs, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| |
Collapse
|
23
|
Sharma D, Zhao F. Updates on clinical trials evaluating the regenerative potential of allogenic mesenchymal stem cells in COVID-19. NPJ Regen Med 2021; 6:37. [PMID: 34193864 PMCID: PMC8245638 DOI: 10.1038/s41536-021-00147-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 06/11/2021] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected nearly 118 million people and caused ~2.6 million deaths worldwide by early 2021, during the coronavirus disease 2019 (COVID-19) pandemic. Although the majority of infected patients show mild-to-moderate symptoms, a small fraction of patients develops severe symptoms. Uncontrolled cytokine production and the lack of substantive adaptive immune response result in hypoxia, acute respiratory distress syndrome (ARDS), or multiple organ failure in severe COVID-19 patients. Since the current standard of care treatment is insufficient to alleviate severe COVID-19 symptoms, many clinics have been prompted to perform clinical trials involving the infusion of mesenchymal stem cells (MSCs) due to their immunomodulatory and therapeutic properties. Several phases I/II clinical trials involving the infusion of allogenic MSCs have been performed last year. The focus of this review is to critically evaluate the safety and efficacy outcomes of the most recent, placebo-controlled phase I/II clinical studies that enrolled a larger number of patients, in order to provide a statistically relevant and comprehensive understanding of MSC's therapeutic potential in severe COVID-19 patients. Clinical outcomes obtained from these studies clearly indicate that: (i) allogenic MSC infusion in COVID-19 patients with ARDS is safe and effective enough to decreases a set of inflammatory cytokines that may drive COVID-19 associated cytokine storm, and (ii) MSC infusion efficiently improves COVID-19 patient survival and reduces recovery time. These findings strongly support further investigation into MSC-infusion in larger clinical trials for COVID-19 patients with ARDS, who currently have a nearly 50% of mortality rate.
Collapse
Affiliation(s)
- Dhavan Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States.
| |
Collapse
|
24
|
Lanzoni G, Linetsky E, Correa D, Messinger Cayetano S, Alvarez RA, Kouroupis D, Alvarez Gil A, Poggioli R, Ruiz P, Marttos AC, Hirani K, Bell CA, Kusack H, Rafkin L, Baidal D, Pastewski A, Gawri K, Leñero C, Mantero AMA, Metalonis SW, Wang X, Roque L, Masters B, Kenyon NS, Ginzburg E, Xu X, Tan J, Caplan AI, Glassberg MK, Alejandro R, Ricordi C. Umbilical cord mesenchymal stem cells for COVID-19 acute respiratory distress syndrome: A double-blind, phase 1/2a, randomized controlled trial. Stem Cells Transl Med 2021; 10:660-673. [PMID: 33400390 PMCID: PMC8046040 DOI: 10.1002/sctm.20-0472] [Citation(s) in RCA: 250] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/22/2020] [Accepted: 12/06/2020] [Indexed: 12/17/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) in COVID-19 is associated with high mortality. Mesenchymal stem cells are known to exert immunomodulatory and anti-inflammatory effects and could yield beneficial effects in COVID-19 ARDS. The objective of this study was to determine safety and explore efficacy of umbilical cord mesenchymal stem cell (UC-MSC) infusions in subjects with COVID-19 ARDS. A double-blind, phase 1/2a, randomized, controlled trial was performed. Randomization and stratification by ARDS severity was used to foster balance among groups. All subjects were analyzed under intention to treat design. Twenty-four subjects were randomized 1:1 to either UC-MSC treatment (n = 12) or the control group (n = 12). Subjects in the UC-MSC treatment group received two intravenous infusions (at day 0 and 3) of 100 ± 20 × 106 UC-MSCs; controls received two infusions of vehicle solution. Both groups received best standard of care. Primary endpoint was safety (adverse events [AEs]) within 6 hours; cardiac arrest or death within 24 hours postinfusion). Secondary endpoints included patient survival at 31 days after the first infusion and time to recovery. No difference was observed between groups in infusion-associated AEs. No serious adverse events (SAEs) were observed related to UC-MSC infusions. UC-MSC infusions in COVID-19 ARDS were found to be safe. Inflammatory cytokines were significantly decreased in UC-MSC-treated subjects at day 6. Treatment was associated with significantly improved patient survival (91% vs 42%, P = .015), SAE-free survival (P = .008), and time to recovery (P = .03). UC-MSC infusions are safe and could be beneficial in treating subjects with COVID-19 ARDS.
Collapse
Affiliation(s)
- Giacomo Lanzoni
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of Biochemistry and Molecular BiologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Elina Linetsky
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Diego Correa
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of Orthopedics, UHealth Sports Medicine InstituteUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Shari Messinger Cayetano
- Division of Biostatistics, Department of Public Health SciencesUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Roger A. Alvarez
- Department of MedicineUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- University of Miami Health SystemMiamiFloridaUSA
| | - Dimitrios Kouroupis
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Ana Alvarez Gil
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Raffaella Poggioli
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Phillip Ruiz
- Department of SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Antonio C. Marttos
- Department of MedicineUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- University of Miami Health SystemMiamiFloridaUSA
- Jackson Health SystemMiamiFloridaUSA
| | - Khemraj Hirani
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of MedicineUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Crystal A. Bell
- Department of MedicineUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Halina Kusack
- Department of MedicineUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Lisa Rafkin
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - David Baidal
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of MedicineUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- University of Miami Health SystemMiamiFloridaUSA
| | | | - Kunal Gawri
- Department of MedicineUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- University of Miami Health SystemMiamiFloridaUSA
| | - Clarissa Leñero
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Alejandro M. A. Mantero
- Division of Biostatistics, Department of Public Health SciencesUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Sarah W. Metalonis
- Division of Biostatistics, Department of Public Health SciencesUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Xiaojing Wang
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Luis Roque
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Burlett Masters
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Norma S. Kenyon
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Enrique Ginzburg
- Department of SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- University of Miami Health SystemMiamiFloridaUSA
- Jackson Health SystemMiamiFloridaUSA
| | - Xiumin Xu
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jianming Tan
- The Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanPeople's Republic of China
| | - Arnold I. Caplan
- Skeletal Research CenterCase Western Reserve UniversityClevelandOhioUSA
| | | | - Rodolfo Alejandro
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of MedicineUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- University of Miami Health SystemMiamiFloridaUSA
| | - Camillo Ricordi
- Diabetes Research Institute, Cell Transplant CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| |
Collapse
|
25
|
Pillalamarri N, Abdullah, Ren G, Khan L, Ullah A, Jonnakuti S, Ullah M. Exploring the utility of extracellular vesicles in ameliorating viral infection-associated inflammation, cytokine storm and tissue damage. Transl Oncol 2021; 14:101095. [PMID: 33887552 PMCID: PMC8053440 DOI: 10.1016/j.tranon.2021.101095] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) have emerged as potential mediators of intercellular communication. EVs are nano-sized, lipid membrane-bound vesicles that contains biological information in the form of proteins, metabolites and/or nucleic acids. EVs are key regulators of tissue repair mechanisms, such as in the context of lung injuries. Recent studies suggest that EVs have the ability to repair COVID19-associated acute lung damage. EVs hold great promise for therapeutic treatments, particularly in treating a potentially fatal autoimmune response and attenuate inflammation. They are known to boost lung immunity and are involved in the pathogenesis of various lung diseases, including viral infection. EV-based immunization technology has been proven to elicit robust immune responses in many models of infectious disease, including COVID-19. The field of EV research has tremendous potential in advancing our understanding about viral infection pathogenesis, and can be translated into anti-viral therapeutic strategies.
Collapse
Affiliation(s)
- Nagavalli Pillalamarri
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA 94304, United States
| | - Abdullah
- Molecular Medicine Department of Medicine, Stanford University, CA, United States
| | - Gang Ren
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA 94304, United States
| | - Luqman Khan
- School of Medicine, University of California, San Francisco, CA 94158, United States
| | - Asad Ullah
- School of Medicine, University of California, San Francisco, CA 94158, United States
| | - Sriya Jonnakuti
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA 94304, United States
| | - Mujib Ullah
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA 94304, United States; Molecular Medicine Department of Medicine, Stanford University, CA, United States.
| |
Collapse
|
26
|
Putra A, Widyatmoko A, Ibrahim S, Amansyah F, Amansyah F, Berlian MA, Retnaningsih R, Pasongka Z, Sari FE, Rachmad B. Case series of the first three severe COVID-19 patients treated with the secretome of hypoxia-mesenchymal stem cells in Indonesia. F1000Res 2021; 10:228. [PMID: 35350705 PMCID: PMC8927740 DOI: 10.12688/f1000research.51191.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2021] [Indexed: 01/21/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the outbreak of coronavirus disease 2019 (COVID-19), which has been rapidly spreading. Several guideline therapies have been proposed as a possible treatment for SARS-CoV-2, however, these therapies are not sufficient to treat a severe condition of SARS-CoV-2 infection characterised by the increase of D-dimer and C-reactive protein (CRP) levels, and patchy ground-glass opacities (GGOs). Secretome-mesenchymal stem cells (S-MSCs) produced by MSCs under hypoxia could excessively release several anti-inflammatory cytokines and growth factors to control the COVID-19 cytokine storm and accelerate lung injury improvement. This is the first study investigating the clinical outcomes of three severe COVID-19 patients admitted to the intensive care unit of three different hospitals in Indonesia treated with S-MSCs. The decrease of D-dimer and CRP level was reported for all patients treated with S-MSCs. This was in line with improvement of pulmonary radiology, blood gas level, and hematologic assessment. In conclusion, these cases suggest that S-MSCs could effectively control D-dimer, CRP level and GGOs of severe COVID-19 patients associated with recovered pulmonary function.
Collapse
Affiliation(s)
- Agung Putra
- Stem Cell and Cancer Research (SCCR), Faculty of Medicine, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia
- Department of Postgraduate Biomedical Science, Faculty of Medicine, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia
- Department of Pathological Anatomy, Faculty of Medicine,, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia
| | - Agus Widyatmoko
- Department of Internal Medicine, Faculty of Medicine, Universitas Muhammadiyah Yogyakarta, Yogyakarta, Yogyakarta, Indonesia
| | - Sugeng Ibrahim
- Stem Cell and Cancer Research (SCCR), Faculty of Medicine, Universitas Islam Sultan Agung (Unissula), Semarang, Central Java, Indonesia
- Department of Molecular Biology, Faculty of Medicine, Universitas Katholik (Unika) Soegijapranata, Semarang, Central Java, Indonesia
| | - Fajar Amansyah
- Department of Internal Medicine, Bhayangkara Hospital, Makassar, South Sulawesi, Indonesia
| | - Farid Amansyah
- Department of Internal Medicine, Bhayangkara Hospital, Makassar, South Sulawesi, Indonesia
| | - Mukti Arja Berlian
- Department of Internal Medicine, Dr. Esnawan Antariksa Air Force Hospital, Jakarta, Jakarta, Indonesia
| | - Retnaningsih Retnaningsih
- Department of Neurology and Intensive Care Unit, Kariadi Hospital, Universitas Diponegoro, Semarang, Central Java, Indonesia
| | | | - Flora Eka Sari
- Department of Pulmonary Medicine, Dr. Esnawan Antariksa Air Force Hospital, Jakarta, Jakarta, Indonesia
| | - Basuki Rachmad
- Department of Intensive Care Unit, Gatot Soebroto Army Hospital, Jakarta, Jakarta, Indonesia
| |
Collapse
|
27
|
Samara A, Herlenius E. Is There an Effect of Fetal Mesenchymal Stem Cells in the Mother-Fetus Dyad in COVID-19 Pregnancies and Vertical Transmission? Front Physiol 2021; 11:624625. [PMID: 33679426 PMCID: PMC7928412 DOI: 10.3389/fphys.2020.624625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/30/2020] [Indexed: 12/15/2022] Open
Abstract
Because of the polysystemic nature of coronavirus disease 2019 (COVID-19), during the present pandemic, there have been serious concerns regarding pregnancy, vertical transmission, and intrapartum risk. The majority of pregnant patients with COVID-19 infection present with mild or asymptomatic course of the disease. Some cases were hospitalized, and few needed intensive care unit admission, or mechanical ventilation. There have also been scarce case reports where neonates required mechanical ventilation post COVID-19 pregnancies. Without approved therapies other than dexamethasone, advanced mesenchymal cell therapy is one immunomodulatory therapeutic approach that is currently explored and might hold great promise. We suggest that the circulating fetal stem cells might have an immune-protective effect to mothers and contribute to the often mild and even asymptomatic post-COVID-19 pregnancies. Thus, COVID-19 pregnancies come forth as a paradigm to be further and more comprehensively approached, to understand both the mechanism and action of circulating stem cells in immunoprotection and hypoxia in microcirculation.
Collapse
Affiliation(s)
- Athina Samara
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- Astrid Lindgren Children′s Hospital Karolinska University Hospital, Stockholm, Sweden
| | - Eric Herlenius
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- Astrid Lindgren Children′s Hospital Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
28
|
Xu X, Jiang W, Chen L, Xu Z, Zhang Q, Zhu M, Ye P, Li H, Yu L, Zhou X, Zhou C, Chen X, Zheng X, Xu K, Cai H, Zheng S, Jiang W, Wu X, Li D, Chen L, Luo Q, Wang Y, Qu J, Li Y, Zheng W, Jiang Y, Tang L, Xiang C, Li L. Evaluation of the safety and efficacy of using human menstrual blood-derived mesenchymal stromal cells in treating severe and critically ill COVID-19 patients: An exploratory clinical trial. Clin Transl Med 2021; 11:e297. [PMID: 33634996 PMCID: PMC7839959 DOI: 10.1002/ctm2.297] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was identified in December 2019 and has subsequently spread worldwide. Currently, there is no effective method to cure COVID-19. Mesenchymal stromal cells (MSCs) may be able to effectively treat COVID-19, especially for severe and critical patients. Menstrual blood-derived MSCs have recently received much attention due to their superior proliferation ability and their lack of ethical problems. Forty-four patients were enrolled from January to April 2020 in a multicenter, open-label, nonrandomized, parallel-controlled exploratory trial. Twenty-six patients received allogeneic, menstrual blood-derived MSC therapy, and concomitant medications (experimental group), and 18 patients received only concomitant medications (control group). The experimental group was treated with three infusions totaling 9 × 107 MSCs, one infusion every other day. Primary and secondary endpoints related to safety and efficacy were assessed at various time points during the 1-month period following MSC infusion. Safety was measured using the frequency of treatment-related adverse events (AEs). Patients in the MSC group showed significantly lower mortality (7.69% died in the experimental group vs 33.33% in the control group; P = .048). There was a significant improvement in dyspnea while undergoing MSC infusion on days 1, 3, and 5. Additionally, SpO2 was significantly improved following MSC infusion, and chest imaging results were improved in the experimental group in the first month after MSC infusion. The incidence of most AEs did not differ between the groups. MSC-based therapy may serve as a promising alternative method for treating severe and critical COVID-19.
Collapse
Affiliation(s)
- Xiaowei Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Wanli Jiang
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanHebeiP.R. China
| | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Zhenyu Xu
- Innovative Precision Medicine (IPM) GroupHangzhouZhejiangP. R. China
| | - Qiang Zhang
- Innovative Precision Medicine (IPM) GroupHangzhouZhejiangP. R. China
| | - Mengfei Zhu
- Shulan (Hangzhou) Hospital, Zhejiang Shuren University Shulan International Medical CollegeHangzhouZhejiangP. R. China
| | - Peng Ye
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanHebeiP.R. China
| | - Hang Li
- Innovative Precision Medicine (IPM) GroupHangzhouZhejiangP. R. China
| | - Liang Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Xiaoyang Zhou
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanHebeiP.R. China
| | - Chenliang Zhou
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanHebeiP.R. China
| | - Xiaobei Chen
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanHebeiP.R. China
| | - Xiaoqin Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Kaijin Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Hongliu Cai
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Shufa Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Wubian Jiang
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanHebeiP.R. China
| | - Xiaojun Wu
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanHebeiP.R. China
| | - Dong Li
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanHebeiP.R. China
| | - Lu Chen
- Innovative Precision Medicine (IPM) GroupHangzhouZhejiangP. R. China
| | - Qingqing Luo
- Innovative Precision Medicine (IPM) GroupHangzhouZhejiangP. R. China
| | - Yingyan Wang
- Innovative Precision Medicine (IPM) GroupHangzhouZhejiangP. R. China
| | - Jingjing Qu
- Department of Respiratory DiseaseThoracic Disease CentreThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yifei Li
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Wendi Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yingan Jiang
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanHebeiP.R. China
| | - Lingling Tang
- Shulan (Hangzhou) Hospital, Zhejiang Shuren University Shulan International Medical CollegeHangzhouZhejiangP. R. China
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Shulan (Hangzhou) Hospital, Zhejiang Shuren University Shulan International Medical CollegeHangzhouZhejiangP. R. China
| |
Collapse
|
29
|
Schultz IC, Bertoni APS, Wink MR. Mesenchymal Stem Cell-Derived Extracellular Vesicles Carrying miRNA as a Potential Multi Target Therapy to COVID-19: an In Silico Analysis. Stem Cell Rev Rep 2021; 17:341-356. [PMID: 33511519 PMCID: PMC7842178 DOI: 10.1007/s12015-021-10122-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2021] [Indexed: 01/08/2023]
Abstract
In the end of 2019 COVID-19 emerged as a new threat worldwide and this disease present impaired immune system, exacerbated production of inflammatory cytokines, and coagulation disturbs. Mesenchymal stem cell (MSC) derived extracellular vesicles (EVs) have emerged as a therapeutic option due to its intrinsic properties to alleviate inflammatory responses, capable to promote the restoring of injured tissue. EVs contain heterogeneous cargo, including active microRNAs, small noncoding sequences involved in post-transcriptional gene repression or degradation and can attach in multiple targets. This study investigated whether the MSC-EVs miRNA cargo has the capacity to modulate the exacerbated cytokines, cell death and coagulation disturbs present in severe COVID-19. Through bioinformatics analysis, four datasets of miRNA, using different stem cell tissue sources (bone marrow, umbilical cord and adipose tissue), and one dataset of mRNA (bone marrow) were analyzed. 58 miRNAs overlap in the four miRNA datasets analyzed. Sequentially, those miRNAs present in at least two datasets, were analyzed using miRWalk for the 3’UTR binding target mRNA. The result predicted 258 miRNAs for exacerbated cytokines and chemokines, 266 miRNAs for cell death genes and 148 miRNAs for coagulation cascades. Some miRNAs may simultaneously attenuate inflammatory agents, inhibit cell death genes and key factors of coagulation cascade, consequently preventing tissue damage and coagulation disturbs. Therefore, the MSC-derived EVs due to their heterogeneous cargo are a potential multitarget approach able to improve the survival rates of severe COVID-19 patients.
Collapse
Affiliation(s)
- Iago Carvalho Schultz
- Laboratório de Biologia Celular, Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, RS, 90050-170, Brazil
| | - Ana Paula Santin Bertoni
- Laboratório de Biologia Celular, Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, RS, 90050-170, Brazil
| | - Márcia Rosângela Wink
- Laboratório de Biologia Celular, Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, RS, 90050-170, Brazil.
| |
Collapse
|
30
|
Ghaffari M, Mollazadeh-Bajestani M, Moztarzadeh F, Uludağ H, Hardy JG, Mozafari M. An overview of the use of biomaterials, nanotechnology, and stem cells for detection and treatment of COVID-19: towards a framework to address future global pandemics. EMERGENT MATERIALS 2021; 4:19-34. [PMID: 33426467 PMCID: PMC7783485 DOI: 10.1007/s42247-020-00143-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/16/2020] [Indexed: 05/03/2023]
Abstract
A novel SARS-like coronavirus (severe acute respiratory syndrome-related coronavirus-2, SARS-CoV-2) outbreak has recently become a worldwide pandemic. Researchers from various disciplinary backgrounds (social to natural science, health and medicine, etc.) have studied different aspects of the pandemic. The current situation has revealed how the ongoing development of nanotechnology and nanomedicine can accelerate the fight against the novel viruses. A comprehensive solution to this and future pandemic outbreaks includes preventing the spread of the virus through anti-viral personal protective equipment (PPE) and anti-viral surfaces, plus efforts to encourage behavior to minimize risks. Studies of previously introduced anti-viral biomaterials and their optimization to fight against SARS-CoV-2 is the foundation of most of the recent progress. The identification of non-symptomatic patients and symptomatic patients is vital. Reviewing published research highlights the pivotal roles of nanotechnology and biomaterials in the development and efficiency of detection techniques, e.g., by applying nanotechnology and nanomedicine as part of the road map in the treatment of coronavirus disease 2019 (COVID-19) patients. In this review, we discuss efforts to deploy nanotechnology, biomaterials, and stem cells in each step of the fight against SARS-CoV-2, which may provide a framework for future efforts in combating global pandemics.
Collapse
Affiliation(s)
- Maryam Ghaffari
- Biomaterials Group, Faculty of Biomedical Engineering (Center of Excellence), Amirkabir University of Technology, Tehran, Iran
| | | | - Fathollah Moztarzadeh
- Biomaterials Group, Faculty of Biomedical Engineering (Center of Excellence), Amirkabir University of Technology, Tehran, Iran
| | - Hasan Uludağ
- Department of Chemical and Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 2V4 Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1 Canada
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3 Canada
| | - John G. Hardy
- Department of Chemistry, Faculty of Science and Technology, Lancaster University, Lancaster, LA1 4YB UK
- Materials Science Institute, Lancaster University, Lancaster, LA1 4YB UK
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Present Address: Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| |
Collapse
|
31
|
Kumar A, Dey AD, Behl T, Chadha S, Aggarwal V. Exploring the multifocal therapeutic approaches in COVID-19: A ray of hope. Int Immunopharmacol 2021; 90:107156. [PMID: 33189613 PMCID: PMC7608010 DOI: 10.1016/j.intimp.2020.107156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/24/2020] [Accepted: 10/21/2020] [Indexed: 12/28/2022]
Abstract
The ongoing global pandemic of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is marked as one of the most challenging infectious diseases in the history of mankind with affliction of ~29,737,453 confirmed cases globally. Looking at the present scenario where there is a parallel increment in curve with time, there is an utmost emergency to discover a perennial solution to this life-threatening virus which has led the Human race in an unusual state of affair. The entire health care fraternity is engaged in endeavouring an ultimate way out to hit this pandemic but no such research made till now has been approved yet, to have the potential to bring an end to this fatal situation. Although a few possible treatment choices exist at the moment yet the requirement to search for a still better therapeutic option remains persistent. Global laboratories are working day and night in search for an effective vaccine, many are undergoing clinical trials but their commercialization is no less than a year away. The present review highlights the current potential therapies viz., vaccines, immunotherapies, convulsant plasma therapies, corticosteroids, antithrombotic, intravenous immunoglobulins, nocturnal oxygen therapy etc. that may prove beneficial in attenuating the pandemic situation. However, comparison and presentation of collective data on the therapeutic advancements in mitigating the pandemic situation needs further clinical investigations in order to prove boon to mankind.
Collapse
Affiliation(s)
- Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Asmita Deka Dey
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Swati Chadha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vishal Aggarwal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
32
|
Ellison-Hughes GM, Colley L, O'Brien KA, Roberts KA, Agbaedeng TA, Ross MD. The Role of MSC Therapy in Attenuating the Damaging Effects of the Cytokine Storm Induced by COVID-19 on the Heart and Cardiovascular System. Front Cardiovasc Med 2020; 7:602183. [PMID: 33363221 PMCID: PMC7756089 DOI: 10.3389/fcvm.2020.602183] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/17/2020] [Indexed: 01/08/2023] Open
Abstract
The global pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes coronavirus disease 2019 (COVID-19) has led to 47 m infected cases and 1. 2 m (2.6%) deaths. A hallmark of more severe cases of SARS-CoV-2 in patients with acute respiratory distress syndrome (ARDS) appears to be a virally-induced over-activation or unregulated response of the immune system, termed a "cytokine storm," featuring elevated levels of pro-inflammatory cytokines such as IL-2, IL-6, IL-7, IL-22, CXCL10, and TNFα. Whilst the lungs are the primary site of infection for SARS-CoV-2, in more severe cases its effects can be detected in multiple organ systems. Indeed, many COVID-19 positive patients develop cardiovascular complications, such as myocardial injury, myocarditis, cardiac arrhythmia, and thromboembolism, which are associated with higher mortality. Drug and cell therapies targeting immunosuppression have been suggested to help combat the cytokine storm. In particular, mesenchymal stromal cells (MSCs), owing to their powerful immunomodulatory ability, have shown promise in early clinical studies to avoid, prevent or attenuate the cytokine storm. In this review, we will discuss the mechanistic underpinnings of the cytokine storm on the cardiovascular system, and how MSCs potentially attenuate the damage caused by the cytokine storm induced by COVID-19. We will also address how MSC transplantation could alleviate the long-term complications seen in some COVID-19 patients, such as improving tissue repair and regeneration.
Collapse
Affiliation(s)
- Georgina M. Ellison-Hughes
- Faculty of Life Sciences & Medicine, Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, King's College London Guy's Campus, London, United Kingdom
| | - Liam Colley
- School of Sport, Health, and Exercise Sciences, Bangor University, Bangor, United Kingdom
| | - Katie A. O'Brien
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Kirsty A. Roberts
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Thomas A. Agbaedeng
- Faculty of Health & Medical Sciences, Centre for Heart Rhythm Disorders, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Mark D. Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| |
Collapse
|
33
|
Amigues I, Pearlman AH, Patel A, Reid P, Robinson PC, Sinha R, Kim AH, Youngstein T, Jayatilleke A, Konigon M. Coronavirus disease 2019: investigational therapies in the prevention and treatment of hyperinflammation. Expert Rev Clin Immunol 2020; 16:1185-1204. [PMID: 33146561 PMCID: PMC7879704 DOI: 10.1080/1744666x.2021.1847084] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
Introduction: The mortality of coronavirus disease 2019 (COVID-19) is frequently driven by an injurious immune response characterized by the development of acute respiratory distress syndrome (ARDS), endotheliitis, coagulopathy, and multi-organ failure. This spectrum of hyperinflammation in COVID-19 is commonly referred to as cytokine storm syndrome (CSS). Areas covered: Medline and Google Scholar were searched up until 15th of August 2020 for relevant literature. Evidence supports a role of dysregulated immune responses in the immunopathogenesis of severe COVID-19. CSS associated with SARS-CoV-2 shows similarities to the exuberant cytokine production in some patients with viral infection (e.g.SARS-CoV-1) and may be confused with other syndromes of hyperinflammation like the cytokine release syndrome (CRS) in CAR-T cell therapy. Interleukin (IL)-6, IL-8, and tumor necrosis factor-alpha have emerged as predictors of COVID-19 severity and in-hospital mortality. Expert opinion: Despite similarities, COVID-19-CSS appears to be distinct from HLH, MAS, and CRS, and the application of HLH diagnostic scores and criteria to COVID-19 is not supported by emerging data. While immunosuppressive therapy with glucocorticoids has shown a mortality benefit, cytokine inhibitors may hold promise as 'rescue therapies' in severe COVID-19. Given the arguably limited benefit in advanced disease, strategies to prevent the development of COVID-19-CSS are needed.
Collapse
Affiliation(s)
- Isabelle Amigues
- Division of Rheumatology, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Alexander H Pearlman
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aarat Patel
- Bon Secours Rheumatology Center and Division of Pediatric Rheumatology, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Pankti Reid
- Division of Rheumatology, Department of Internal Medicine, Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago Medical Center, Chicago, IL, USA
| | - Philip C. Robinson
- School of Clinical Medicine, University of Queensland Faculty of Medicine, Queensland, Australia
| | - Rashmi Sinha
- Department of Medicine, Systemic Juvenile Idiopathic Arthritis Foundation, Cincinnati, OH, USA
| | - Alfred Hj Kim
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
- Andrew M. And Jane M. Bursky Center of Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
| | - Taryn Youngstein
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Arundathi Jayatilleke
- Division of Rheumatology, Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Maximilian Konigon
- Division of Rheumatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
34
|
Li Z, Niu S, Guo B, Gao T, Wang L, Wang Y, Wang L, Tan Y, Wu J, Hao J. Stem cell therapy for COVID-19, ARDS and pulmonary fibrosis. Cell Prolif 2020; 53:e12939. [PMID: 33098357 PMCID: PMC7645923 DOI: 10.1111/cpr.12939] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an acute respiratory infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 mainly causes damage to the lung, as well as other organs and systems such as the hearts, the immune system and so on. Although the pathogenesis of COVID-19 has been fully elucidated, there is no specific therapy for the disease at present, and most treatments are limited to supportive care. Stem cell therapy may be a potential treatment for refractory and unmanageable pulmonary illnesses, which has shown some promising results in preclinical studies. In this review, we systematically summarize the pathogenic progression and potential mechanisms underlying stem cell therapy in COVID-19, and registered COVID-19 clinical trials. Of all the stem cell therapies touted for COVID-19 treatment, mesenchymal stem cells (MSCs) or MSC-like derivatives have been the most promising in preclinical studies and clinical trials so far. MSCs have been suggested to ameliorate the cytokine release syndrome (CRS) and protect alveolar epithelial cells by secreting many kinds of factors, demonstrating safety and possible efficacy in COVID-19 patients with acute respiratory distress syndrome (ARDS). However, considering the consistency and uniformity of stem cell quality cannot be quantified nor guaranteed at this point, more work remains to be done in the future.
Collapse
Affiliation(s)
- Zhongwen Li
- Institute of ZoologyState Key Laboratory of Stem Cell and Reproductive BiologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- National Stem Cell Resource CenterChinese Academy of SciencesBeijingChina
| | - Shuaishuai Niu
- Institute of ZoologyState Key Laboratory of Stem Cell and Reproductive BiologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- National Stem Cell Resource CenterChinese Academy of SciencesBeijingChina
| | - Baojie Guo
- Institute of ZoologyState Key Laboratory of Stem Cell and Reproductive BiologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- National Stem Cell Resource CenterChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Tingting Gao
- Institute of ZoologyState Key Laboratory of Stem Cell and Reproductive BiologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- National Stem Cell Resource CenterChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Lei Wang
- Institute of ZoologyState Key Laboratory of Stem Cell and Reproductive BiologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- National Stem Cell Resource CenterChinese Academy of SciencesBeijingChina
| | - Yukai Wang
- Institute of ZoologyState Key Laboratory of Stem Cell and Reproductive BiologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- National Stem Cell Resource CenterChinese Academy of SciencesBeijingChina
| | - Liu Wang
- Institute of ZoologyState Key Laboratory of Stem Cell and Reproductive BiologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- National Stem Cell Resource CenterChinese Academy of SciencesBeijingChina
| | - Yuanqing Tan
- Institute of ZoologyState Key Laboratory of Stem Cell and Reproductive BiologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- National Stem Cell Resource CenterChinese Academy of SciencesBeijingChina
| | - Jun Wu
- Institute of ZoologyState Key Laboratory of Stem Cell and Reproductive BiologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- National Stem Cell Resource CenterChinese Academy of SciencesBeijingChina
| | - Jie Hao
- Institute of ZoologyState Key Laboratory of Stem Cell and Reproductive BiologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- National Stem Cell Resource CenterChinese Academy of SciencesBeijingChina
| |
Collapse
|
35
|
Can A, Coskun H. The rationale of using mesenchymal stem cells in patients with COVID-19-related acute respiratory distress syndrome: What to expect. Stem Cells Transl Med 2020; 9:1287-1302. [PMID: 32779878 PMCID: PMC7404450 DOI: 10.1002/sctm.20-0164] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/06/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2)‐caused coronavirus disease 2019 (COVID‐19) pandemic has become a global health crisis with an extremely rapid progress resulting in thousands of patients who may develop acute respiratory distress syndrome (ARDS) requiring intensive care unit (ICU) treatment. So far, no specific antiviral therapeutic agent has been demonstrated to be effective for COVID‐19; therefore, the clinical management is largely supportive and depends on the patients' immune response leading to a cytokine storm followed by lung edema, dysfunction of air exchange, and ARDS, which could lead to multiorgan failure and death. Given that human mesenchymal stem cells (MSCs) from various tissue sources have revealed successful clinical outcomes in many immunocompromised disorders by inhibiting the overactivation of the immune system and promoting endogenous repair by improving the microenvironment, there is a growing demand for MSC infusions in patients with COVID‐19‐related ARDS in the ICU. In this review, we have documented the rationale and possible outcomes of compassionate use of MSCs, particularly in patients with SARS‐CoV‐2 infections, toward proving or disproving the efficacy of this approach in the near future. Many centers have registered and approved, and some already started, single‐case or phase I/II trials primarily aiming to rescue their critical patients when no other therapeutic approach responds. On the other hand, it is also very important to mention that there is a good deal of concern about clinics offering unproven stem cell treatments for COVID‐19. The reviewers and oversight bodies will be looking for a balanced but critical appraisal of current trials.
Collapse
Affiliation(s)
- Alp Can
- Laboratory for Stem Cells and Reproductive Cell Biology, Department of Histology and Embryology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Hakan Coskun
- Harvard Medical School, Boston, Massachusetts, USA.,Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|